1
|
Qian Z, Lin W, Cai X, Wu J, Ke K, Ye Z, Wu F. WYC-209 inhibited GC malignant progression by down-regulating WNT4 through RARα. Cancer Biol Ther 2024; 25:2299288. [PMID: 38178596 PMCID: PMC10773637 DOI: 10.1080/15384047.2023.2299288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
Gastric cancer (GC) has been a major health burden all over the world but there are fewer promising chemotherapeutic drugs due to its multidrug resistance. It has been reported that WYC-209 suppresses the growth and metastasis of tumor-repopulating cells but the effect on GC was not explored. MTT, colony formation, and transwell assays were performed to examine the effects of WYC-209 on the proliferation, colony growth, and mobility of GC cells. Western blotting and qRT-PCR were used to detect the expression of proteins and mRNA. RNA-seq and enrichment analyses were conducted for the differentially expressed genes and enriched biological processes and pathways. The rescue experiments were carried out for further validation. Besides, we constructed xenograft model to confirm the effect of WYC-209 in vivo. The dual-luciferase reporter and Chromatin immunoprecipitation were implemented to confirm the underlying mechanism. WYC-209 exerted excellent anti-cancer effects both in vitro and in vivo. Based on RNA-seq and enrichment analyses, we found that Wnt family member 4 (WNT4) was significantly down-regulated. More importantly, WNT4 overexpression breached the inhibitory effect of WYC-209 on GC progression. Mechanically, WYC-209 significantly promoted the binding between retinoic acid receptor α (RARα) and WNT4 promoter. WYC-209 exerts anti-tumor effects in GC by down-regulating the expression of WNT4 via RARα.
Collapse
Affiliation(s)
- Zhenyuan Qian
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenfa Lin
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xufan Cai
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianzhang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zaiyuan Ye
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Qian Z, Lin W, Cai X, Wu J, Ke K, Ye Z, Wu F. WYC-209 suppresses gastric cancer by down-regulating FGF18 via inactivating the STAT3 signaling pathway. Eur J Pharmacol 2024; 983:176957. [PMID: 39214271 DOI: 10.1016/j.ejphar.2024.176957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Gastric cancer (GC) is regarded as a major health burden all over the world. WYC-209 inhibits the growth and metastasis of tumor-repopulating cells (TRCs). However, its effectiveness on GC was unexplored. Herein, this study aims to investigate the effect of WYC-209 on GC and elucidate its underlying mechanism. METHODS We examined the effects of WYC-209 on cell survival, migration, invasion, and colony-forming capacities of two GC cell lines (AGS and HGC-27). Subsequently, RNA-seq and enrichment analyses were performed to screen the differentially expressed genes (DEGs) and the enriched signaling pathways. To further explore the underlying mechanism, loss- and gain-function experiments, Chromatin immunoprecipitation, and luciferase reporter were conducted. Finally, xenograft models were constructed to examine the effects of WYC-209 in vivo. RESULTS WYC-209 significantly inhibited cell motility in vitro and tumor growth in vivo. RNA-seq performed in AGS cells after WYC-209 treatment revealed that the inhibition effect of WYC-209 on GCs may be associated with the down-regulation of fibroblast growth factor-18 (FGF18), and pleasantly, FGF18 overexpression abrogated the suppression effect of the drug. In addition, we found that WYC-209 attenuated the activation of the Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway, and impeded the FGF18 levels expressed in GCs. Importantly, the WYC-209 treatment circumvented the binding of STAT3 to the FGF18 promoter, suggested that WYC-209 down-regulated FGF18 expression via the STAT3 signaling pathway. CONCLUSION Together, our findings presented the promise of WYC-209 in suppressing GC by down-regulating FGF18 expression through inactivating the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhenyuan Qian
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenfa Lin
- Zhejiang Chinese Medical University, China
| | - Xufan Cai
- Zhejiang Chinese Medical University, China
| | - Jianzhang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zaiyuan Ye
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Fang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Jun SY, Hong SM, Jang KT. Prognostic Significance of Cyclin D1 Expression in Small Intestinal Adenocarcinoma. Cancers (Basel) 2023; 15:5032. [PMID: 37894399 PMCID: PMC10604933 DOI: 10.3390/cancers15205032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Cyclin D1, a critical cyclin-dependent kinase (CDK) 4/6-dependent regulator of G1/S transition, has attracted much interest as a therapeutic target. The cyclin D1 expression in small intestinal adenocarcinomas (SIACs) has not yet been comprehensively studied, owing to the rarity of this tumor. We investigated the clinicopathological and prognostic significance of the cyclin D1 expression in 232 surgically resected primary SIACs through a multi-institutional study. A high expression of cyclin D1 (cyclin D1High) was detected in 145 SIAC cases (63%), which was significantly higher than that in normal small intestinal mucosa (11%). Cyclin D1High was more commonly found in SIACs with a lower T-category and disease stage and KRAS mutation and predicted better patient survival. Multivariate analysis revealed that cyclin D1High, the absence of retroperitoneal seeding and lymphovascular invasion, and the lower N-category were identified as independent prognostic indicators for patients with SIACs. Specifically, cyclin D1High affected patient survival in the lower stage group (stages I and II). In conclusion, cyclin D1 was commonly overexpressed in SIACs, and cyclin D1High acted as a favorable prognostic indicator in patients with SIACs. These findings in SIACs may, thus, be important to further comprehend the mechanism of cyclin D1 in carcinogenesis and to strategize appropriate patient therapies.
Collapse
Affiliation(s)
- Sun-Young Jun
- Department of Pathology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Republic of Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Kee-Taek Jang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| |
Collapse
|
4
|
Wei CH, Huang L, Kreh B, Liu X, Tyutyunyk-Massey L, Kawakami M, Chen Z, Shi M, Kozlov S, Chan KC, Andresson T, Carrington M, Vuligonda V, Sanders ME, Horowitz A, Hwu P, Peng W, Dmitrovsky E, Liu X. A novel retinoic acid receptor-γ agonist antagonizes immune checkpoint resistance in lung cancers by altering the tumor immune microenvironment. Sci Rep 2023; 13:14907. [PMID: 37689790 PMCID: PMC10492813 DOI: 10.1038/s41598-023-41690-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
All-trans-retinoic acid (ATRA), the retinoic acid receptors (RARs) agonist, regulates cell growth, differentiation, immunity, and survival. We report that ATRA-treatment repressed cancer growth in syngeneic immunocompetent, but not immunodeficient mice. The tumor microenvironment was implicated: CD8+ T cell depletion antagonized ATRA's anti-tumorigenic effects in syngeneic mice. ATRA-treatment with checkpoint blockade did not cooperatively inhibit murine lung cancer growth. To augment ATRA's anti-tumorigenicity without promoting its pro-tumorigenic potential, an RARγ agonist (IRX4647) was used since it regulates T cell biology. Treating with IRX4647 in combination with an immune checkpoint (anti-PD-L1) inhibitor resulted in a statistically significant suppression of syngeneic 344SQ lung cancers in mice-a model known for its resistance to checkpoints and characterized by low basal T cell and PD-L1 expression. This combined treatment notably elevated CD4+ T-cell presence within the tumor microenvironment and increased IL-5 and IL-13 tumor levels, while simultaneously decreasing CD38 in the tumor stroma. IL-5 and/or IL-13 treatments increased CD4+ more than CD8+ T-cells in mice. IRX4647-treatment did not appreciably affect in vitro lung cancer growth, despite RARγ expression. Pharmacokinetic analysis found IRX4647 plasma half-life was 6 h in mice. Yet, RARα antagonist (IRX6696)-treatment with anti-PD-L1 did not repress syngeneic lung cancer growth. Together, these findings provide a rationale for a clinical trial investigating an RARγ agonist to augment check point blockade response in cancers.
Collapse
Affiliation(s)
- Cheng-Hsin Wei
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Lu Huang
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Blair Kreh
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Xiuxia Liu
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Liliya Tyutyunyk-Massey
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Masanori Kawakami
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Zibo Chen
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Mi Shi
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Serguei Kozlov
- Center for Advanced Preclinical Research, Frederick, MD, USA
| | - King C Chan
- Protein Characterization Laboratory, Frederick, MD, USA
| | | | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Amir Horowitz
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Moffitt Cancer Center, Tampa, FL, USA
| | - Weiyi Peng
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan Dmitrovsky
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA
| | - Xi Liu
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, PO Box B, Frederick, MD, 21701, USA.
| |
Collapse
|
5
|
Bexarotene-induced cell death in ovarian cancer cells through Caspase-4-gasdermin E mediated pyroptosis. Sci Rep 2022; 12:11123. [PMID: 35778597 PMCID: PMC9249775 DOI: 10.1038/s41598-022-15348-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/22/2022] [Indexed: 11/08/2022] Open
Abstract
Bexarotene selectively activates retinoid X receptor, which is a commonly used anticancer agent for cutaneous T-cell lymphoma. In this study, we aimed to investigate the anticancer effect of bexarotene and its underlying mechanism in ovarian cancer in vitro. The ES2 and NIH:OVACAR3 ovarian cancer cell lines were treated with 0, 5, 10, or 20 µM of bexarotene. After 24 h, cell number measurement and lactate dehydrogenase (LDH) cytotoxicity assay were performed. The effect of bexarotene on CDKN1A expression, cell cycle-related protein, cell cycle, pyroptosis, and apoptosis was evaluated. Bexarotene reduced cell proliferation in all concentrations in both the cells. At concentrations of > 10 µM, extracellular LDH activity increased with cell rupture. Treatment using 10 µM of bexarotene increased CDKN1A mRNA levels, decreased cell cycle-related protein expression, and increased the sub-G1 cell population in both cells. In ES2 cells, caspase-4 and GSDME were activated, whereas caspase-3 was not, indicating that bexarotene-induced cell death might be pyroptosis. A clinical setting concentration of bexarotene induced cell death through caspase-4-mediated pyroptosis in ovarian cancer cell lines. Thus, bexarotene may serve as a novel therapeutic agent for ovarian cancer.
Collapse
|
6
|
Deligiorgi MV, Trafalis DT. The continuum of care of anticancer treatment-induced hypothyroidism in patients with solid non thyroid tumors: time for an intimate collaboration between oncologists and endocrinologists. Expert Rev Clin Pharmacol 2022; 15:531-549. [PMID: 35757870 DOI: 10.1080/17512433.2022.2093714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hypothyroidism is a common adverse event of various anticancer treatment modalities, constituting a notable paradigm of the integration of the endocrine perspective into precision oncology. AREAS COVERED The present narrative review provides a comprehensive and updated overview of anticancer treatment-induced hypothyroidism in patients with solid non-thyroid tumors. A study search was conducted on the following electronic databases: PubMed, Google Scholar, Scopus.com, ClinicalTrials.gov, and European Union Clinical Trials Register from 2011 until August 2021. EXPERT OPINION In patients with solid non-thyroid tumors, hypothyroidism is a common adverse event of radiotherapy, high dose interleukin 2 (HD IL-2), interferon alpha (IFN-α), bexarotene, immune checkpoint inhibitors (ICPi), and tyrosine kinase inhibitors (TKIs), while chemotherapy may induce hypothyroidism more often than initially considered. The path forward for the management of anticancer treatment-induced hypothyroidism in patients with solid non-thyroid tumors is an integrated approach grounded on 5 pillars: prevention, vigilance, diagnosis, treatment and monitoring. Current challenges concerning anticancer treatment-induced hypothyroidism await counteraction, namely awareness of the growing list of related anticancer treatments, identification of predictive factors, counteraction of diagnostic pitfalls, tuning of thyroid hormone replacement, and elucidation of its prognostic significance. Close collaboration of oncologists with endocrinologists will provide optimal patient care.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| |
Collapse
|
7
|
Leal AS, Reich LA, Moerland JA, Zhang D, Liby KT. Potential therapeutic uses of rexinoids. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 91:141-183. [PMID: 34099107 DOI: 10.1016/bs.apha.2021.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The discovery of nuclear receptors, particularly retinoid X receptors (RXR), and their involvement in numerous pathways related to development sparked interest in their immunomodulatory properties. Genetic models using deletion or overexpression of RXR and the subsequent development of several small molecules that are agonists or antagonists of this receptor support a promising therapeutic role for these receptors in immunology. Bexarotene was approved in 1999 for the treatment of cutaneous T cell lymphoma. Several other small molecule RXR agonists have since been synthesized with limited preclinical development, but none have yet achieved FDA approval. Cancer treatment has recently been revolutionized with the introduction of immune checkpoint inhibitors, but their success has been restricted to a minority of patients. This review showcases the emerging immunomodulatory effects of RXR and the potential of small molecules that target this receptor as therapies for cancer and other diseases. Here we describe the essential roles that RXR and partner receptors play in T cells, dendritic cells, macrophages and epithelial cells, especially within the tumor microenvironment. Most of these effects are site and cancer type dependent but skew immune cells toward an anti-inflammatory and anti-tumor effect. This beneficial effect on immune cells supports the promise of combining rexinoids with approved checkpoint blockade therapies in order to enhance efficacy of the latter and to delay or potentially eliminate drug resistance. The data compiled in this review strongly suggest that targeting RXR nuclear receptors is a promising new avenue in immunomodulation for cancer and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ana S Leal
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Lyndsey A Reich
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jessica A Moerland
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Di Zhang
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Karen T Liby
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
8
|
Xia L, Shen D, Zhang Y, Lu J, Wang M, Wang H, Chen Y, Xue D, Xie D, Li G. Targeting the TR4 nuclear receptor with antagonist bexarotene can suppress the proopiomelanocortin signalling in AtT-20 cells. J Cell Mol Med 2021; 25:2404-2417. [PMID: 33491272 PMCID: PMC7933964 DOI: 10.1111/jcmm.16074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/29/2020] [Accepted: 10/25/2020] [Indexed: 01/12/2023] Open
Abstract
Drug options for the life‐threatening Cushing's disease are limited, and surgical resection or radiation therapy is not invariably effective. Testicular receptor 4 (TR4) has been identified as a novel drug target to treat Cushing's disease. We built the structure model of TR4 and searched the TR4 antagonist candidate via in silico virtual screening. Bexarotene was identified as an antagonist of TR4 that can directly interact with TR4 ligand binding domain (TR4‐LBD) and induces a conformational change in the secondary structure of TR4‐LBD. Bexarotene suppressed AtT‐20 cell growth, proopiomelanocortin (POMC) expression and adrenocorticotropin (ACTH) secretion. Mechanism dissection revealed that bexarotene could suppress TR4‐increased POMC expression via promoting the TR4 translocation from the nucleus to the cytoplasm. This TR4 translocation might then result in reducing the TR4 binding to the TR4 response element (TR4RE) on the 5’ promoter region of POMC. Results from in vivo mouse model also revealed that oral bexarotene administration markedly suppressed ACTH‐secreting tumour growth, adrenal enlargement and the secretion of ACTH and corticosterone in mice with already established tumours. Together, these results suggest that bexarotene may be developed as a potential novel therapeutic drug to better suppress Cushing's disease.
Collapse
Affiliation(s)
- Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danyang Shen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Youyun Zhang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jieyang Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingchao Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huan Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanlei Chen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingwei Xue
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dajiang Xie
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Moerland JA, Zhang D, Reich LA, Carapellucci S, Lockwood B, Leal AS, Krieger-Burke T, Aleiwi B, Ellsworth E, Liby KT. The novel rexinoid MSU-42011 is effective for the treatment of preclinical Kras-driven lung cancer. Sci Rep 2020; 10:22244. [PMID: 33335263 PMCID: PMC7746742 DOI: 10.1038/s41598-020-79260-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Effective drugs are needed for lung cancer, as this disease remains the leading cause of cancer-related deaths. Rexinoids are promising drug candidates for cancer therapy because of their ability to modulate genes involved in inflammation, cell proliferation or differentiation, and apoptosis through activation of the retinoid X receptor (RXR). The only currently FDA-approved rexinoid, bexarotene, is ineffective as a single agent for treating epithelial cancers and induces hypertriglyceridemia. Here, we used a previously validated screening paradigm to evaluate 23 novel rexinoids for biomarkers related to efficacy and safety. These biomarkers include suppression of inducible nitric oxide synthase (iNOS) and induction of sterol regulatory element-binding protein (SREBP). Because of its potent iNOS suppression, low SREBP induction, and activation of RXR, MSU-42011 was selected as our lead compound. We next used MSU-42011 to treat established tumors in a clinically relevant Kras-driven mouse model of lung cancer. KRAS is one of the most common driver mutations in human lung cancer and correlates with aggressive disease progression and poor patient prognosis. Ultrasound imaging was used to detect and monitor tumor development and growth over time in the lungs of the A/J mice. MSU-42011 markedly decreased the tumor number, size, and histopathology of lung tumors compared to the control and bexarotene groups. Histological sections of lung tumors in mice treated with MSU-42011 exhibited reduced cell density and fewer actively proliferating cells compared to the control and bexarotene-treated tumors. Although bexarotene significantly (p < 0.01) elevated plasma triglycerides and cholesterol, treatment with MSU-42011 did not increase these biomarkers, demonstrating a more favorable toxicity profile in vivo. The combination of MSU-42011 and carboplatin and paclitaxel reduced macrophages in the lung and increased activation markers of CD8+T cells compared to the control groups. Our results validate our screening paradigm for in vitro testing of novel rexinoids and demonstrate the potential for MSU-42011 to be developed for the treatment of KRAS-driven lung cancer.
Collapse
Affiliation(s)
- Jessica A Moerland
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Di Zhang
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Lyndsey A Reich
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Sarah Carapellucci
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Beth Lockwood
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Ana S Leal
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
| | - Teresa Krieger-Burke
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
- In Vivo Facility, Michigan State University, East Lansing, MI, USA
| | - Bilal Aleiwi
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
- Medicial Chemistry Core, Michigan State University, East Lansing, MI, USA
| | - Edmund Ellsworth
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA
- Medicial Chemistry Core, Michigan State University, East Lansing, MI, USA
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI, 48824, USA.
| |
Collapse
|
10
|
Dong Z, Wang Y, Ding V, Yan X, Lv Y, Zhong M, Zhu F, Zhao P, He C, Ding F, Shi H. GLI1 activation is a key mechanism of erlotinib resistance in human non-small cell lung cancer. Oncol Lett 2020; 20:76. [PMID: 32863909 PMCID: PMC7436900 DOI: 10.3892/ol.2020.11937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 06/23/2020] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-associated death worldwide. In recent years, the advancement of epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) targeted therapies has provided clinical benefits for lung cancer patients with EGFR mutations. The response to EGFR-TKI varies in patients with lung cancer, and resistance typically develops during the course of the treatment. Therefore, understanding biomarkers which can predict resistance to EGFR-TKI is important. Overexpression of GLI causes activation of the Hedgehog (Hh) signaling pathway and plays a critical role in oncogenesis in numerous types of cancer. In the present study, the role of GLI1 in erlotinib resistance was investigated. GLI1 mRNA and protein expression levels were determined using reverse transcription-quantitative PCR and immunohistochemistry (IHC) in lung cancer cell lines and tumor specimens, respectively. GLI1 mRNA expression levels were found to be positively correlated with the IC50 of erlotinib in 15 non-small cell lung cancer (NSCLC) cell lines. The downregulation of GLI1 using siRNA sensitized lung cancer cells to the erlotinib treatment, whereas the overexpression of GLI1 increased the survival of lung cancer cells in the presence of erlotinib, indicating that Hh/GLI activation may play a critical role in the development of TKI resistance in lung cancer. Combined treatment with erlotinib and a GLI1 inhibitor reduced the cell viability synergistically. A retrospective study of patients with NSCLC treated with erlotinib revealed that those with a high IHC score for GLI1 protein expression had a poorer prognosis. These results indicated that GLI1 is a key regulator for TKI sensitivity, and patients with lung cancer may benefit from the combined treatment of TKI and GLI1 inhibitor.
Collapse
Affiliation(s)
- Zhouhuan Dong
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yun Wang
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Vivianne Ding
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Xiang Yan
- Department of Medical Oncology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Yali Lv
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Mei Zhong
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Fengwei Zhu
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Po Zhao
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Charlotte He
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Feng Ding
- Zhejiang Provincial Key Laboratory of Applied Enzymology and Precision Medicine Center, Jiaxing, Zhejiang 314006, P.R. China.,ACCB Diagnostic Laboratory, Yangze Delta Region Institute of Tsinghua University Zhejiang, Jiaxing, Zhejiang 314006, P.R. China
| | - Huaiyin Shi
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
11
|
He H, Xu C, Cheng Z, Qian X, Zheng L. Drug Combinatorial Therapies for the Treatment of KRAS Mutated Lung Cancers. Curr Top Med Chem 2019; 19:2128-2142. [PMID: 31475900 DOI: 10.2174/1568026619666190902150555] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/23/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
KRAS is the most common oncogene to be mutated in lung cancer, and therapeutics directly targeting KRAS have proven to be challenging. The mutations of KRAS are associated with poor prognosis, and resistance to both adjuvant therapy and targeted EGFR TKI. EGFR TKIs provide significant clinical benefit for patients whose tumors bear EGFR mutations. However, tumors with KRAS mutations rarely respond to the EGFR TKI therapy. Thus, combination therapy is essential for the treatment of lung cancers with KRAS mutations. EGFR TKI combined with inhibitors of MAPKs, PI3K/mTOR, HDAC, Wee1, PARP, CDK and Hsp90, even miRNAs and immunotherapy, were reviewed. Although the effects of the combination vary, the combined therapeutics are one of the best options at present to treat KRAS mutant lung cancer.
Collapse
Affiliation(s)
- Hao He
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Chang Xu
- National Vaccine & Serum Institute, Beijing, China
| | - Zhao Cheng
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xiaoying Qian
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lei Zheng
- School of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Shastri S, Chatterjee B, Thakur SS. Achievements in Cancer Research and its Therapeutics in Hundred Years. Curr Top Med Chem 2019; 19:1545-1562. [PMID: 31362690 DOI: 10.2174/1568026619666190730093034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
Abstract
Cancer research has progressed leaps and bounds over the years. This review is a brief overview of the cancer research, milestone achievements and therapeutic studies on it over the one hundred ten years which would give us an insight into how far we have come to understand and combat this fatal disease leading to millions of deaths worldwide. Modern biology has proved that cancer is a very complex disease as still we do not know precisely how it triggers. It involves several factors such as protooncogene, oncogene, kinase, tumor suppressor gene, growth factor, signalling cascade, micro RNA, immunity, environmental factors and carcinogens. However, modern technology now helps the cancer patient on the basis of acquired and established knowledge in the last hundred years to save human lives.
Collapse
Affiliation(s)
- Sravanthi Shastri
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Bhaswati Chatterjee
- National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Suman S Thakur
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
13
|
Mohammed A, Fox JT, Miller MS. Cancer Chemoprevention: Preclinical In Vivo Alternate Dosing Strategies to Reduce Drug Toxicities. Toxicol Sci 2019; 170:251-259. [PMID: 31020311 PMCID: PMC6657562 DOI: 10.1093/toxsci/kfz104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer chemopreventive agents inhibit the formation of precursor lesions and/or the progression of these lesions to late stage disease. This approach to disease control has the potential to reduce the physical and financial costs of cancer in society. Several drugs that have been approved by the FDA for other diseases and have been extensively evaluated for their safety and pharmacokinetic/pharmacodynamic characteristics have the potential to be repurposed for use as cancer chemopreventive agents. These agents often mechanistically inhibit signaling molecules that play key roles in the carcinogenic process. The safety profile of agents is a primary concern when considering the administration of drugs for chemoprevention, as the drugs will be given chronically to high-risk, asymptomatic individuals. To decrease drug toxicity while retaining efficacy, several approaches are currently being explored. In this short review, we describe studies that use preclinical in vivo models to assess efficacy of alternative drug dosing strategies and routes of drug administration on chemopreventive drug efficacy. In vivo drug dosing strategies that reduce toxicity while retaining efficacy will pave the way for future cancer prevention clinical trials.
Collapse
Affiliation(s)
- Altaf Mohammed
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Jennifer T Fox
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Mark Steven Miller
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
14
|
Zhang D, Leal AS, Carapellucci S, Shahani PH, Bhogal JS, Ibrahim S, Raban S, Jurutka PW, Marshall PA, Sporn MB, Wagner CE, Liby KT. Testing Novel Pyrimidinyl Rexinoids: A New Paradigm for Evaluating Rexinoids for Cancer Prevention. Cancer Prev Res (Phila) 2019; 12:211-224. [PMID: 30760500 DOI: 10.1158/1940-6207.capr-18-0317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/10/2018] [Accepted: 02/04/2019] [Indexed: 11/16/2022]
Abstract
Rexinoids, selective ligands for retinoid X receptors (RXR), have shown promise in preventing many types of cancer. However, the limited efficacy and undesirable lipidemic side-effects of the only clinically approved rexinoid, bexarotene, drive the search for new and better rexinoids. Here we report the evaluation of novel pyrimidinyl (Py) analogues of two known chemopreventive rexinoids, bexarotene (Bex) and LG100268 (LG268) in a new paradigm. We show that these novel derivatives were more effective agents than bexarotene for preventing lung carcinogenesis induced by a carcinogen. In addition, these new analogues have an improved safety profile. PyBex caused less elevation of plasma triglyceride levels than bexarotene, while PyLG268 reduced plasma cholesterol levels and hepatomegaly compared with LG100268. Notably, this new paradigm mechanistically emphasizes the immunomodulatory and anti-inflammatory activities of rexinoids. We reveal new immunomodulatory actions of the above rexinoids, especially their ability to diminish the percentage of macrophages and myeloid-derived suppressor cells in the lung and to redirect activation of M2 macrophages. The rexinoids also potently inhibit critical inflammatory mediators including IL6, IL1β, CCL9, and nitric oxide synthase (iNOS) induced by lipopolysaccharide. Moreover, in vitro iNOS and SREBP (sterol regulatory element-binding protein) induction assays correlate with in vivo efficacy and toxicity, respectively. Our results not only report novel pyrimidine derivatives of existing rexinoids, but also describe a series of biological screening assays that will guide the synthesis of additional rexinoids. Further progress in rexinoid synthesis, potency, and safety should eventually lead to a clinically acceptable and useful new drug for patients with cancer.
Collapse
Affiliation(s)
- Di Zhang
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Ana S Leal
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Sarah Carapellucci
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Pritika H Shahani
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Jaskaran S Bhogal
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Samir Ibrahim
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - San Raban
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Peter W Jurutka
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Pamela A Marshall
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Michael B Sporn
- Geisel School of Medicine at Dartmouth, Department of Pharmacology, Lebanon, New Hampshire
| | - Carl E Wagner
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, Arizona
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
15
|
Dragnev KH, Whyman JD, Hahn CK, Kebbekus PE, Kokko SF, Bhatt SM, Rigas JR. A phase I/II study of bexarotene with carboplatin and weekly paclitaxel for the treatment of patients with advanced non-small cell lung cancer. J Thorac Dis 2018; 10:5531-5537. [PMID: 30416803 PMCID: PMC6196173 DOI: 10.21037/jtd.2018.09.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/30/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Rexinoids demonstrate anti-proliferative differentiation-inducing activity in multiple cancer types, including NSCLC. Prior studies have shown promising results when combining rexinoids with chemotherapy. This phase I/II study evaluates the tolerability and activity of a rexinoid, bexarotene, combined with weekly paclitaxel and monthly carboplatin. METHODS Patients with confirmed advanced stage IIIB or IV NSCLC and adequate organ function were enrolled. They were scheduled to receive carboplatin (AUC =6) and 3 doses of weekly paclitaxel (100 mg/m2) every 4 weeks. Oral bexarotene was administered daily at two doses: 300 and 400 mg/m2/day. RESULTS Thirty-three patients were enrolled. Fourteen received 300 mg/m2/day and 19 received 400 mg/m2/day of bexarotene. Hematologic toxicity included grade 3 neutropenia in 7 patients. Hyperlipidemia was a major non-hematologic toxicity which was medically managed. The recommended phase II dose of bexarotene was 400 mg/m2/day. Response rate was 35%. Median overall survival (OS) for all patients was 8.3 months with 1-year survival of 43%. Median OS for the 300 mg/m2 dose of bexarotene was 6.6 versus 9.8 months for the 400 mg/m2 dose (HR, 0.73; Log rank P=0.37). Patients who experienced hypertriglyceridemia had a median OS of 9.8 months compared to 4.9 months for those who did not (HR, 0.69; Log rank P=0.33). CONCLUSIONS The 43% 1-year survival for patients receiving bexarotene with weekly paclitaxel and monthly carboplatin is encouraging. With the availability of new classes of agents for lung cancer, further evaluation of this regimen in unselected patients is not warranted. Our study confirms prior subgroup analyses showing a significant correlation between bexarotene-induced hypertriglyceridemia and survival. Further research is needed to identify molecular biomarkers to identify this subset of patients and to explore rexinoids in other combinations, especially with immunotherapy.
Collapse
Affiliation(s)
- Konstantin H Dragnev
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- The Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Jeremy D Whyman
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Cynthia K Hahn
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Peter E Kebbekus
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Sarah F Kokko
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Sunil M Bhatt
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - James R Rigas
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- The Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
16
|
Shen D, Yu X, Wu Y, Chen Y, Li G, Cheng F, Xia L. Emerging roles of bexarotene in the prevention, treatment and anti-drug resistance of cancers. Expert Rev Anticancer Ther 2018. [PMID: 29521139 DOI: 10.1080/14737140.2018.1449648] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Danyang Shen
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoming Yu
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Wu
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanlei Chen
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gonghui Li
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Cheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liqun Xia
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Ooi A, Oyama T, Nakamura R, Tajiri R, Ikeda H, Fushida S, Dobashi Y. Gene amplification of CCNE1, CCND1, and CDK6 in gastric cancers detected by multiplex ligation-dependent probe amplification and fluorescence in situ hybridization. Hum Pathol 2016; 61:58-67. [PMID: 27864121 DOI: 10.1016/j.humpath.2016.10.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/19/2016] [Accepted: 10/28/2016] [Indexed: 12/18/2022]
Abstract
New and effective treatments for advanced gastric cancer are urgently needed. Cyclins E and D1 form a complex with cyclin-dependent kinase 2, 4, or 6 to regulate G1-S transition. The G1-S regulatory genes encoding cyclin E (CCNE1), cyclin D1 (CCND1), and CDK6 (CDK6) are frequently amplified in gastric cancer and may therefore influence molecularly targeted therapies against ERBB2 or EGFR when coamplified. A total of 179 formalin-fixed and paraffin-embedded gastric cancer specimens were examined for these gene amplifications by multiplex ligation-dependent probe amplification and fluorescence in situ hybridization. Amplification of at least 1 G1-S regulatory gene was found in 35 tumors (CCNE1 amplification, 15% of samples; CCND1, 6%; CDK6, 1%). In 13 of the 35 tumors, dual-color fluorescence in situ hybridization identified coamplification of the G1-S regulatory genes with ERBB2, EGFR, and/or KRAS in single cancer nuclei. The observation that cells with G1-S regulatory gene amplification contained clonal subpopulations with coamplification of ERBB2, EGFR, or KRAS in 5 early and 3 advanced cancers suggests that amplification of the G1-S regulatory genes represents an early event, which precedes ERBB2, EGFR, or KRAS amplification. Amplified CCNE1, CCND1, and CDK6 in advanced gastric cancer may be potentially useful as direct targets for molecular therapy or for combination therapy with ERBB2 or EGFR inhibitors. Multiplex ligation-dependent probe amplification could be a useful tool for identification of patients who would benefit from such therapies.
Collapse
Affiliation(s)
- Akishi Ooi
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa 920-8641, Japan; Pathology Section, University Hospital, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Takeru Oyama
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Ritsuko Nakamura
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Ryousuke Tajiri
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Hiroko Ikeda
- Pathology Section, University Hospital, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa 920-8641, Japan.
| | - Yoh Dobashi
- Department of Pathology, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan.
| |
Collapse
|
18
|
Marchwicka A, Cunningham A, Marcinkowska E, Brown G. Therapeutic use of selective synthetic ligands for retinoic acid receptors: a patent review. Expert Opin Ther Pat 2016; 26:957-71. [PMID: 27336223 DOI: 10.1080/13543776.2016.1205586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Differentiation therapy using all-trans retinoic acid (ATRA) revolutionised the treatment of acute promyelocytic leukaemia to such an extent that it is now one of the most curable types of leukaemia, with ATRA and anthracycline-based chemotherapy providing cure rates above 80%. Isotretinoin is used to treat chronic acne. Here, we examine the information described in recent patents and the extent to which new findings are influencing extending retinoid-based differentiation therapy to other cancers, as well as the development of new therapies for other disorders. AREAS COVERED A search has been performed on the literature and worldwide patents filed during 2014 to the present time, focusing on synthetic agonists and antagonists of retinoic acid receptors and novel compositions for the delivery of these agents. EXPERT OPINION New potential therapeutic applications have been described, including lung, breast and head and neck cancers, T cell lymphoma and neurodegenerative, metabolic, ophthalmic, muscle, and inflammatory disorders. Recent patents have described the means to maximise retinoid activity. Two decades of efforts to extend retinoid-based therapies have been disappointing and new synthetic retinoids, target diseases and modes of delivery may well resolve this long standing issue.
Collapse
Affiliation(s)
- Aleksandra Marchwicka
- a Laboratory of Protein Biochemistry, Faculty of Biotechnology , University of Wroclaw , Wroclaw , Poland
| | - Alan Cunningham
- b Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK
| | - Ewa Marcinkowska
- a Laboratory of Protein Biochemistry, Faculty of Biotechnology , University of Wroclaw , Wroclaw , Poland
| | - Geoffrey Brown
- c Institute of Clinical Sciences, College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK
| |
Collapse
|
19
|
Retinoid X Receptor Agonists Upregulate Genes Responsible for the Biosynthesis of All-Trans-Retinoic Acid in Human Epidermis. PLoS One 2016; 11:e0153556. [PMID: 27078158 PMCID: PMC4831765 DOI: 10.1371/journal.pone.0153556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/31/2016] [Indexed: 11/26/2022] Open
Abstract
UAB30 is an RXR selective agonist that has been shown to have potential cancer chemopreventive properties. Due to high efficacy and low toxicity, it is currently being evaluated in human Phase I clinical trials by the National Cancer Institute. While UAB30 shows promise as a low toxicity chemopreventive drug, the mechanism of its action is not well understood. In this study, we investigated the effects of UAB30 on gene expression in human organotypic skin raft cultures and mouse epidermis. The results of this study indicate that treatment with UAB30 results in upregulation of genes responsible for the uptake and metabolism of all-trans-retinol to all-trans-retinoic acid (ATRA), the natural agonist of RAR nuclear receptors. Consistent with the increased expression of these genes, the steady-state levels of ATRA are elevated in human skin rafts. In ultraviolet B (UVB) irradiated mouse skin, the expression of ATRA target genes is found to be reduced. A reduced expression of ATRA sensitive genes is also observed in epidermis of mouse models of UVB-induced squamous cell carcinoma and basal cell carcinomas. However, treatment of mouse skin with UAB30 prior to UVB irradiation prevents the UVB-induced decrease in expression of some of the ATRA-responsive genes. Considering its positive effects on ATRA signaling in the epidermis and its low toxicity, UAB30 could be used as a chemoprophylactic agent in the treatment of non-melanoma skin cancer, particularly in organ transplant recipients and other high risk populations.
Collapse
|
20
|
Abstract
Carotenoids are polyenes synthesized in plants and certain microorganisms and are pigments used by plants and animals in various physiological processes. Some of the over 600 known carotenoids are capable of metabolic conversion to the essential nutrient vitamin A (retinol) in higher animals. Vitamin A also gives rise to a number of other metabolites which, along with their analogs, are known as retinoids. To facilitate discussion about these important molecules, a nomenclature is required to identify specific substances. The generally accepted rules for naming these important molecules have been agreed to by various Commissions of the International Union of Pure and Applied Chemistry and International Union of Biochemistry. These naming conventions are explained along with comparisons to more systematic naming rules that apply for these organic chemicals. Identification of the carotenoids and retinoids has been advanced by their chemical syntheses, and here, both classical and modern methods for synthesis of these molecules, as well as their analogs, are described. Because of their importance in biological systems, sensitive methods for the detection and quantification of these compounds from various sources have been essential. Early analyses that relied on liquid adsorption and partition chromatography have given way to high-performance liquid chromatography (HPLC) coupled with various detection methods. The development of HPLC coupled to mass spectrometry, particularly LC/MS-MS with Multiple Reaction Monitoring, has resulted in the greatest sensitivity and specificity in these analyses.
Collapse
Affiliation(s)
- Earl H Harrison
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA.
| | - Robert W Curley
- College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
21
|
The Rexinoids LG100268 and LG101506 Inhibit Inflammation and Suppress Lung Carcinogenesis in A/J Mice. Cancer Prev Res (Phila) 2015; 9:105-14. [DOI: 10.1158/1940-6207.capr-15-0325] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/03/2015] [Indexed: 11/16/2022]
|
22
|
Abstract
Early in the age of modern medicine the consequences of vitamin A deficiency drew attention to the fundamental link between retinoid-dependent homeostatic regulation and malignant hyperproliferative diseases. The term "retinoid" includes a handful of endogenous and a large group of synthetic derivatives of vitamin A. These multifunctional lipid-soluble compounds directly regulate target genes of specific biological functions and critical signaling pathways to orchestrate complex functions from vision to development, metabolism, and inflammation. Many of the retinoid activities on the cellular level have been well characterized and translated to the regulation of processes like differentiation and cell death, which play critical roles in the outcome of malignant transformation of tissues. In fact, retinoid-based differentiation therapy of acute promyelocytic leukemia was one of the first successful examples of molecularly targeted treatment strategies. The selectivity, high receptor binding affinity and the ability of retinoids to directly modulate gene expression programs present a distinct pharmacological opportunity for cancer treatment and prevention. However, to fully exploit their potential, the adverse effects of retinoids must be averted. In this review we provide an overview of the biology of retinoid (activated by nuclear retinoic acid receptors [RARs]) and rexinoid (engaged by nuclear retinoid X receptors [RXRs]) action concluded from a long line of preclinical studies, in relation to normal and transformed states of cells. We will also discuss the past and current uses of retinoids in the treatment of malignancies, the potential of rexinoids in the cancer prevention setting, both as single agents and in combinations.
Collapse
Affiliation(s)
- Iván P Uray
- Department of Clinical Cancer Prevention, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Ethan Dmitrovsky
- Department of Clinical Cancer Prevention, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
Oyama T, Okamoto K, Nakamura R, Tajiri R, Ikeda H, Ninomiya I, Ooi A. Overexpression and gene amplification of both ERBB2 and EGFR in an esophageal squamous cell carcinoma revealed by fluorescence in situ hybridization, multiplex ligation-dependent probe amplification and immunohistochemistry. Pathol Int 2015; 65:608-13. [PMID: 26314265 DOI: 10.1111/pin.12344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022]
Abstract
EGFR and ERBB2 belong to the EGFR gene family. In esophageal squamous cell carcinomas (SCCs), amplification of EGFR or ERBB2 is usually mutually exclusive. EGFR amplification occurs in approximately 15% of SCCs, ERBB2 occurs in less than 5%. Here, we report the co-amplification of EGFR and ERBB2 in an ulcerative and infiltrating-type SCC that measured approximately 4.2 × 2.7 × 1.2 cm with a superficial lesion occurring in the thoracic esophagus of a 72-year-old man. Multiplex ligation-dependent probe amplification using representative tumor sections showed gain of CCND1 and coincident amplification of ERBB2 or EGFR or neither. Immunohistochemistry and fluorescence in situ hybridization revealed that the tumor comprised three cancer-cell populations: well-differentiated SCC with high-level ERBB2 amplification and ERBB2 overexpression, more infiltrative poorly-differentiated SCC with high-level EGFR amplification and EGFR overexpression, and poorly-differentiated SCC lacking any ERBB2 or EGFR abnormality. These three populations each had low-level CCND1 amplification and nuclear cyclin D1 overexpression. This histological topology and gene amplification combinations suggested that genetic instability first produced CCND1 amplification, and then ERBB2 or EGFR gene amplification occurred. It is further speculated that during cancer progression and clonal selection indecisive predominance of either clone caused the rare co-amplification of ERBB2 and EGFR in a single chimeric tumor.
Collapse
Affiliation(s)
- Takeru Oyama
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Koichi Okamoto
- Department of Gastroenterologic Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Ritsuko Nakamura
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Ryosuke Tajiri
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Hiroko Ikeda
- Pathology Section, University Hospital, Kanazawa University, Ishikawa, Japan
| | - Itasu Ninomiya
- Department of Gastroenterologic Surgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Akishi Ooi
- Department of Molecular and Cellular Pathology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan.,Pathology Section, University Hospital, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
24
|
Hu S, Lu Y, Orr B, Godek K, Mustachio LM, Kawakami M, Sekula D, Compton DA, Freemantle S, Dmitrovsky E. Specific CP110 Phosphorylation Sites Mediate Anaphase Catastrophe after CDK2 Inhibition: Evidence for Cooperation with USP33 Knockdown. Mol Cancer Ther 2015; 14:2576-85. [PMID: 26304236 DOI: 10.1158/1535-7163.mct-15-0443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/19/2015] [Indexed: 11/16/2022]
Abstract
Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCF(cyclinF), which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non-small cell lung cancers.
Collapse
Affiliation(s)
- Shanhu Hu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Yun Lu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Kristina Godek
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Lisa Maria Mustachio
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Masanori Kawakami
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David Sekula
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Duane A Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Sarah Freemantle
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
25
|
Hu S, Danilov AV, Godek K, Orr B, Tafe LJ, Rodriguez-Canales J, Behrens C, Mino B, Moran CA, Memoli VA, Mustachio LM, Galimberti F, Ravi S, DeCastro A, Lu Y, Sekula D, Andrew AS, Wistuba II, Freemantle S, Compton DA, Dmitrovsky E. CDK2 Inhibition Causes Anaphase Catastrophe in Lung Cancer through the Centrosomal Protein CP110. Cancer Res 2015; 75:2029-38. [PMID: 25808870 DOI: 10.1158/0008-5472.can-14-1494] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 02/13/2015] [Indexed: 01/06/2023]
Abstract
Aneuploidy is frequently detected in human cancers and is implicated in carcinogenesis. Pharmacologic targeting of aneuploidy is an attractive therapeutic strategy, as this would preferentially eliminate malignant over normal cells. We previously discovered that CDK2 inhibition causes lung cancer cells with more than two centrosomes to undergo multipolar cell division leading to apoptosis, defined as anaphase catastrophe. Cells with activating KRAS mutations were especially sensitive to CDK2 inhibition. Mechanisms of CDK2-mediated anaphase catastrophe and how activated KRAS enhances this effect were investigated. Live-cell imaging provided direct evidence that following CDK2 inhibition, lung cancer cells develop multipolar anaphase and undergo multipolar cell division with the resulting progeny apoptotic. The siRNA-mediated repression of the CDK2 target and centrosome protein CP110 induced anaphase catastrophe of lung cancer cells. In contrast, CP110 overexpression antagonized CDK2 inhibitor-mediated anaphase catastrophe. Furthermore, activated KRAS mutations sensitized lung cancer cells to CDK2 inhibition by deregulating CP110 expression. Thus, CP110 is a critical mediator of CDK2 inhibition-driven anaphase catastrophe. Independent examination of murine and human paired normal-malignant lung tissues revealed marked upregulation of CP110 in malignant versus normal lung. Human lung cancers with KRAS mutations had significantly lower CP110 expression as compared with KRAS wild-type cancers. Thus, a direct link was found between CP110 and CDK2 inhibitor antineoplastic response. CP110 plays a mechanistic role in response of lung cancer cells to CDK2 inhibition, especially in the presence of activated KRAS mutations.
Collapse
Affiliation(s)
- Shanhu Hu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Alexey V Danilov
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Kristina Godek
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Laura J Tafe
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Department of Pathology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carmen Behrens
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara Mino
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cesar A Moran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vincent A Memoli
- The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Department of Pathology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Lisa Maria Mustachio
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Fabrizio Galimberti
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Saranya Ravi
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Andrew DeCastro
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Yun Lu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - David Sekula
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Angeline S Andrew
- The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah Freemantle
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Duane A Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
26
|
Tang XH, Osei-Sarfo K, Urvalek AM, Zhang T, Scognamiglio T, Gudas LJ. Combination of bexarotene and the retinoid CD1530 reduces murine oral-cavity carcinogenesis induced by the carcinogen 4-nitroquinoline 1-oxide. Proc Natl Acad Sci U S A 2014; 111:8907-12. [PMID: 24927566 PMCID: PMC4066471 DOI: 10.1073/pnas.1404828111] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We investigated the effects of bexarotene (a retinoid X receptor agonist), CD1530 (a retinoic acid receptor γ selective agonist), and the combination of these two drugs for the prevention of oral carcinogenesis induced by the carcinogen 4-nitroquinoline 1-oxide (4-NQO) in a mouse model of human oral-cavity and esophageal squamous-cell carcinoma previously generated in our laboratory. We observed decreased numbers of neoplastic tongue lesions and reduced lesion severity in the 4-NQO plus CD1530 (4N+C) and 4-NQO plus bexarotene plus CD1530 (4N+B+C) groups compared with the 4-NQO group. RNA-Seq analyses showed increases in transcripts in cell proliferation/cell cycle progression pathways in the 4-NQO vs. the untreated group. In addition, β-catenin and matrix metallopeptidase 9 (MMP9) protein levels and reactive oxygen species (ROS), as assessed by 4-hydroxynonenal (4-HNE) staining, were elevated in tongue tissues 17 wk after the termination of the 4-NQO treatment. The 4N+B, 4N+C, and 4N+B+C groups showed dramatically lower levels of β-catenin, MMP9, and 4-HNE staining compared with the 4-NQO group. The major reduction in 4-HNE staining in the retinoid treatment groups suggests a novel mechanism of action, reduction of ROS, by which bexarotene and CD1530 inhibit carcinogenesis.
Collapse
Affiliation(s)
| | | | | | - Tuo Zhang
- Genomics Resources Core Facility, and
| | | | | |
Collapse
|
27
|
Busch AM, Galimberti F, Nehls KE, Roengvoraphoj M, Sekula D, Li B, Guo Y, Direnzo J, Fiering SN, Spinella MJ, Robbins DJ, Memoli VA, Freemantle SJ, Dmitrovsky E. All-trans-retinoic acid antagonizes the Hedgehog pathway by inducing patched. Cancer Biol Ther 2014; 15:463-72. [PMID: 24496080 DOI: 10.4161/cbt.27821] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Male germ cell tumors (GCTs) are a model for a curable solid tumor. GCTs can differentiate into mature teratomas. Embryonal carcinomas (ECs) represent the stem cell compartment of GCTs and are the malignant counterpart to embryonic stem (ES) cells. GCTs and EC cells are useful to investigate differentiation therapy and chemotherapy response. This study explored mechanistic interactions between all-trans-retinoic acid (RA), which induces differentiation of EC and ES cells, and the Hedgehog (Hh) pathway, a regulator of self-renewal and proliferation. RA was found to induce mRNA and protein expression of Patched 1 (Ptch1), the Hh ligand receptor and negative regulator of this pathway. PTCH1 is also a target gene of Hh signaling through Smoothened (Smo) activation. Yet, this observed RA-mediated Ptch1 induction was independent of Smo. It occurred despite co-treatment with RA and Smo inhibitors. Retinoid induction of Ptch1 also occurred in other RA-responsive cancer cell lines and in normal ES cells. Notably, this enhanced Ptch1 expression was preceded by induction of the homeobox transcription factor Meis1, a direct RA target. Direct interaction between Meis1 and Ptch1 was confirmed using chromatin immunoprecipitation assays. To establish the translational relevance of this work, Ptch1 expression was shown to be deregulated in human ECs relative to mature teratoma and the normal seminiferous tubule. Taken together, these findings reveal a previously unrecognized mechanism through which RA can inhibit the Hh pathway via Ptch1 induction. Engaging this pathway is a new way to repress the Hh pathway that can be translated into the cancer clinic.
Collapse
Affiliation(s)
- Alexander M Busch
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - Fabrizio Galimberti
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | | | - Monic Roengvoraphoj
- Department of Medicine; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA
| | - David Sekula
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - Bin Li
- Molecular Oncology Program; Department of Surgery; Miller School of Medicine; University of Miami; Miami, FL USA
| | - Yongli Guo
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - James Direnzo
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA
| | - Steven N Fiering
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA; Department of Immunology and Microbiology; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Department of Genetics; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - Michael J Spinella
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA
| | - David J Robbins
- Molecular Oncology Program; Department of Surgery; Miller School of Medicine; University of Miami; Miami, FL USA
| | - Vincent A Memoli
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA; Department of Pathology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - Sarah J Freemantle
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Department of Medicine; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth; Hanover, NH USA; Dartmouth-Hitchcock Medical Center; Lebanon, NH USA
| |
Collapse
|
28
|
Phase I and pharmacokinetic study of bexarotene in combination with gefitinib in the third-line treatment of non-small-cell lung cancer: brief report. Anticancer Drugs 2014; 24:731-5. [PMID: 23552470 DOI: 10.1097/cad.0b013e32836100d7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gefitinib (an epidermal growth factor receptor tyrosine kinase inhibitor) and bexarotene (a rexinoid) affect similar oncogenic pathways and are both metabolized through cytochrome P450 CYP3A4. We studied the combination of bexarotene and gefitinib in the third-line treatment of advanced non-small-cell lung cancer to examine pharmacokinetic interactions and establish the maximum tolerated dose. This was a single-institution, nonrandomized, open-label, phase I clinical trial with a standard 3+3 dose escalation. Three patients were enrolled at each dose level on the basis of pharmacokinetic analysis with dose level 1 including gefitinib (Iressa) 250 mg oral daily and bexarotene (Targretin) 400 mg/m oral daily and dose level +1 including gefitinib 500 mg oral daily and bexarotene 400 mg/m oral daily. Patients received gefitinib alone for 2 weeks to allow for steady state and thereafter, bexarotene was added. In dose level 1, two of three patients had undetectable gefitinib levels at day 15 for unknown reasons. However, the peak levels on day 29 for all three patients receiving 250 mg of gefitinib with bexarotene are lower than published peak levels. Among the three patients in dose level +1, ∼40% lower gefitinib plasma concentrations were noted on day 29 compared with day 15 along with a mean 44% reduction in area under the plasma concentration-time curve from 0 to 24 h (AUC0-24). Bexarotene appears to lower the C max and AUC0-24 of gefitinib through cytochrome P450 CYP3A4. Our results have pharmacokinetic implications for ongoing trials that combine bexarotene with other small molecules in the era of personalized cancer therapy.
Collapse
|
29
|
Yim CY, Mao P, Spinella MJ. Headway and hurdles in the clinical development of dietary phytochemicals for cancer therapy and prevention: lessons learned from vitamin A derivatives. AAPS JOURNAL 2014; 16:281-8. [PMID: 24431081 DOI: 10.1208/s12248-014-9562-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 01/11/2023]
Abstract
Accumulating epidemiologic and preclinical evidence support the pharmacologic use of a variety of dietary chemicals for the prevention and treatment of cancer. However, it will be challenging to translate these findings into routine clinical practice since phytochemicals have pleiotropic biological activities that have to be balanced for optimal efficacy without unacceptable and potentially unanticipated toxicities. Correctly matching patient populations and settings with optimal, natural product-based phytochemical therapies will require a greater understanding of the specific mechanisms underlying the efficacy, toxicity, and resistance of each agent in a variety of normal, premalignant, and malignant settings. This, in turn, necessitates continued commitment from the basic research community to guide carefully designed and informed clinical trials. The most developed class of anticancer phytochemicals consists of the derivatives of vitamin A called retinoids. Unlike other natural product chemicals currently under study, the retinoids have been extensively tested in humans. Over 30 years of clinical investigation has resulted in several disappointments, but there were some spectacular successes where certain retinoid-based protocols are now FDA-approved standard of care therapies to treat specific malignancies. Furthermore, retinoids are one of the most evaluated pharmacologic agents in the ultra-challenging setting of interventional cancer prevention. This review will summarize the development of retinoids in cancer therapy and prevention with an emphasis on currently proposed mechanisms mediating their efficacy, toxicity, and resistance.
Collapse
Affiliation(s)
- Christina Y Yim
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, New Hampshire, 03755, USA
| | | | | |
Collapse
|
30
|
Ma T, Galimberti F, Erkmen CP, Memoli V, Chinyengetere F, Sempere L, Beumer JH, Anyang BN, Nugent W, Johnstone D, Tsongalis GJ, Kurie JM, Li H, Direnzo J, Guo Y, Freemantle SJ, Dragnev KH, Dmitrovsky E. Comparing histone deacetylase inhibitor responses in genetically engineered mouse lung cancer models and a window of opportunity trial in patients with lung cancer. Mol Cancer Ther 2013; 12:1545-55. [PMID: 23686769 DOI: 10.1158/1535-7163.mct-12-0933] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Histone deacetylase inhibitor (HDACi; vorinostat) responses were studied in murine and human lung cancer cell lines and genetically engineered mouse lung cancer models. Findings were compared with a window of opportunity trial in aerodigestive tract cancers. In human (HOP62, H522, and H23) and murine transgenic (ED-1, ED-2, LKR-13, and 393P, driven, respectively, by cyclin E, degradation-resistant cyclin E, KRAS, or KRAS/p53) lung cancer cell lines, vorinostat reduced growth, cyclin D1, and cyclin E levels, but induced p27, histone acetylation, and apoptosis. Other biomarkers also changed. Findings from transgenic murine lung cancer models were integrated with those from a window of opportunity trial that measured vorinostat pharmacodynamic responses in pre- versus posttreatment tumor biopsies. Vorinostat repressed cyclin D1 and cyclin E expression in murine transgenic lung cancers and significantly reduced lung cancers in syngeneic mice. Vorinostat also reduced cyclin D1 and cyclin E expression, but increased p27 levels in post- versus pretreatment human lung cancer biopsies. Notably, necrotic and inflammatory responses appeared in posttreatment biopsies. These depended on intratumoral HDACi levels. Therefore, HDACi treatments of murine genetically engineered lung cancer models exert similar responses (growth inhibition and changes in gene expression) as observed in lung cancer cell lines. Moreover, enhanced pharmacodynamic responses occurred in the window of opportunity trial, providing additional markers of response that can be evaluated in subsequent HDACi trials. Thus, combining murine and human HDACi trials is a strategy to translate preclinical HDACi treatment outcomes into the clinic. This study uncovered clinically tractable mechanisms to engage in future HDACi trials.
Collapse
Affiliation(s)
- Tian Ma
- Departments of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Busch AM, Johnson KC, Stan RV, Sanglikar A, Ahmed Y, Dmitrovsky E, Freemantle SJ. Evidence for tankyrases as antineoplastic targets in lung cancer. BMC Cancer 2013; 13:211. [PMID: 23621985 PMCID: PMC3644501 DOI: 10.1186/1471-2407-13-211] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/19/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND New pharmacologic targets are urgently needed to treat or prevent lung cancer, the most common cause of cancer death for men and women. This study identified one such target. This is the canonical Wnt signaling pathway, which is deregulated in cancers, including those lacking adenomatous polyposis coli or β-catenin mutations. Two poly-ADP-ribose polymerase (PARP) enzymes regulate canonical Wnt activity: tankyrase (TNKS) 1 and TNKS2. These enzymes poly-ADP-ribosylate (PARsylate) and destabilize axin, a key component of the β-catenin phosphorylation complex. METHODS This study used comprehensive gene profiles to uncover deregulation of the Wnt pathway in murine transgenic and human lung cancers, relative to normal lung. Antineoplastic consequences of genetic and pharmacologic targeting of TNKS in murine and human lung cancer cell lines were explored, and validated in vivo in mice by implantation of murine transgenic lung cancer cells engineered with reduced TNKS expression relative to controls. RESULTS Microarray analyses comparing Wnt pathway members in malignant versus normal tissues of a murine transgenic cyclin E lung cancer model revealed deregulation of Wnt pathway components, including TNKS1 and TNKS2. Real-time PCR assays independently confirmed these results in paired normal-malignant murine and human lung tissues. Individual treatments of a panel of human and murine lung cancer cell lines with the TNKS inhibitors XAV939 and IWR-1 dose-dependently repressed cell growth and increased cellular axin 1 and tankyrase levels. These inhibitors also repressed expression of a Wnt-responsive luciferase construct, implicating the Wnt pathway in conferring these antineoplastic effects. Individual or combined knockdown of TNKS1 and TNKS2 with siRNAs or shRNAs reduced lung cancer cell growth, stabilized axin, and repressed tumor formation in murine xenograft and syngeneic lung cancer models. CONCLUSIONS Findings reported here uncovered deregulation of specific components of the Wnt pathway in both human and murine lung cancer models. Repressing TNKS activity through either genetic or pharmacological approaches antagonized canonical Wnt signaling, reduced murine and human lung cancer cell line growth, and decreased tumor formation in mouse models. Taken together, these findings implicate the use of TNKS inhibitors to target the Wnt pathway to combat lung cancer.
Collapse
Affiliation(s)
- Alexander M Busch
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Wang S, Peng L, Li J, Zeng X, Ouyang L, Tan C, Lu Q. A trial-based cost-effectiveness analysis of erlotinib alone versus platinum-based doublet chemotherapy as first-line therapy for Eastern Asian nonsquamous non-small-cell lung cancer. PLoS One 2013; 8:e55917. [PMID: 23520448 PMCID: PMC3592875 DOI: 10.1371/journal.pone.0055917] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 01/03/2013] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Lung cancer, the most prevalent malignant cancer in the world, remains a serious threat to public health. Recently, a large number of studies have shown that an epidermoid growth factor receptor-tyrosine kinase inhibitor (EGFR TKI), Erlotinib, has significantly better efficacy and is better tolerated in advanced non-small cell lung cancer (NSCLC) patients with a positive EGFR gene mutation. However, access to this drug is severely limited in China due to its high acquisition cost. Therefore, we decided to conduct a study to compare cost-effectiveness between erlotinib monotherapy and carboplatin-gemcitabine (CG) combination therapy in patients with advanced EGFR mutation-positive NSCLC. METHODS A Markov model was developed from the perspective of the Chinese health care system to evaluate the cost-effectiveness of the two treatment strategies; this model was based on data from the OPTIMAL trial, which was undertaken at 22 centres in China. The 10-year quality-adjusted life years (QALYs), direct costs, and incremental cost-effectiveness ratio (ICER) were estimated. To allow for uncertainties within the parameters and to estimate the model robustness, one-way sensitivity analysis and probabilistic sensitivity analysis were performed. RESULTS The median progression-free survival (PFS) obtained from Markov model was 13.2 months (13.1 months was reported in the trial) in the erlotinib group while and 4.64 months (4.6 months was reported in the trial) in the CG group. The QALYs were 1.4 years in the erlotinib group and 1.96 years in the CG group, indicating difference of 0.56 years. The ICER was most sensitive to the health utility of DP ranged from $58,584.57 to $336,404.2. At a threshold of $96,884, erlotinib had a 50% probability of being cost-effective. CONCLUSIONS Erlotinib monotherapy is more cost-effective compared with platinum-based doublets chemotherapy as a first-line therapy for advanced EGFR mutation- positive NSCLC patients from within the Chinese health care system.
Collapse
Affiliation(s)
- Siying Wang
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- School of Pharmaceutical Sciences, Central South University, Changsha Hunan, China
| | - Liubao Peng
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- * E-mail:
| | - Jianhe Li
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- School of Pharmaceutical Sciences, Central South University, Changsha Hunan, China
| | - Xiaohui Zeng
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- School of Pharmaceutical Sciences, Central South University, Changsha Hunan, China
| | - Lihui Ouyang
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- School of Pharmaceutical Sciences, Central South University, Changsha Hunan, China
| | - Chongqing Tan
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
- School of Pharmaceutical Sciences, Central South University, Changsha Hunan, China
| | - Qiong Lu
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha Hunan, China
| |
Collapse
|
34
|
Vedell PT, Lu Y, Grubbs CJ, Yin Y, Jiang H, Bland KI, Muccio DD, Cvetkovic D, You M, Lubet R. Effects on gene expression in rat liver after administration of RXR agonists: UAB30, 4-methyl-UAB30, and Targretin (Bexarotene). Mol Pharmacol 2013; 83:698-708. [PMID: 23292798 DOI: 10.1124/mol.112.082404] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Examination of three retinoid X receptor (RXR) agonists [Targretin (TRG), UAB30, and 4-methyl-UAB30 (4-Me-UAB30)] showed that all inhibited mammary cancer in rodents and two (TRG and 4-Me-UAB30) strikingly increased serum triglyceride levels. Agents were administered in diets to female Sprague-Dawley rats. Liver RNA was isolated and microarrayed on the Affymetrix GeneChip Rat Exon 1.0 ST array. Statistical tests identified genes that exhibited differential expression and fell into groups, or modules, with differential expression among agonists. Genes in specific modules were changed by one, two, or all three agonists. An interactome analysis assessed the effects on genes that heterodimerize with known nuclear receptors. For proliferator-activated receptor α/RXR-activated genes, the strongest response was TRG > 4-Me-UAB30 > UAB30. Many liver X receptor/RXR-related genes (e.g., Scd-1 and Srebf1, which are associated with increased triglycerides) were highly expressed in TRG and 4-Me-UAB30- but not UAB30-treated livers. Minimal expression changes were associated with retinoic acid receptor or vitamin D receptor heterodimers by any of the agonists. UAB30 unexpectedly and uniquely activated genes associated with the aryl hydrocarbon hydroxylase (Ah) receptor (Cyp1a1, Cyp1a2, Cyp1b1, and Nqo1). Based on the Ah receptor activation, UAB30 was tested for its ability to prevent dimethylbenzanthracene (DMBA)-induced mammary cancers, presumably by inhibiting DMBA activation, and was highly effective. Gene expression changes were determined by reverse transcriptase-polymerase chain reaction in rat livers treated with Targretin for 2.3, 7, and 21 days. These showed similar gene expression changes at all three time points, arguing some steady-state effect. Different patterns of gene expression among the agonists provided insight into molecular differences and allowed one to predict certain physiologic consequences of agonist treatment.
Collapse
Affiliation(s)
- Peter T Vedell
- Medical College of Wisconsin, Cancer Center, Department of Pharmacology Toxicology, Milwaukee, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gold KA, Kim ES, Wistuba II, Hong WK. Personalizing lung cancer prevention through a reverse migration strategy. Top Curr Chem (Cham) 2013; 329:221-40. [PMID: 22752582 PMCID: PMC3737590 DOI: 10.1007/128_2012_338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung cancer is the deadliest cancer in the United States and worldwide. Tobacco use is the one of the primary causes of lung cancer and smoking cessation is an important step towards prevention, but patients who have quit smoking remain at risk for lung cancer. Finding pharmacologic agents to prevent lung cancer could potentially save many lives. Unfortunately, despite extensive research, there are no known effective chemoprevention agents for lung cancer. Clinical trials in the past, using agents without a clear target in an unselected population, have shown pharmacologic interventions to be ineffective or even harmful. We propose a new approach to drug development in the chemoprevention setting: reverse migration, that is, drawing on our experience in the treatment of advanced cancer to bring agents, biomarkers, and study designs into the prevention setting. By identifying molecular drivers of lung neoplasia and using matched targeted agents, we hope to personalize therapy to each individual to develop more effective, tolerable chemoprevention. Also, advances in risk modeling, using not only clinical characteristics but also biomarkers, may help us to select patients better for chemoprevention efforts, thus sparing patients at low risk for cancer the potential toxicities of treatment. Our institution has experience with biomarker-driven clinical trials, as in the recently reported Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) trial, and we now propose to bring this trial design into the prevention setting.
Collapse
Affiliation(s)
- Kathryn A Gold
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
36
|
Dragnev K, You M, Wang Y, Lubet R. Lung cancer chemoprevention: difficulties, promise and potential agents? Expert Opin Investig Drugs 2012; 22:35-47. [PMID: 23167766 DOI: 10.1517/13543784.2013.731392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION In a variety of cancers there is evidence that specific regimens can prevent or significantly delay the development of cancer. Thus, for breast cancer (ER+) use of SERMs or aromatase inhibitors can substantially decrease tumor incidence. For cervical cancer, HPV vaccination will inhibit long term cancer incidence. For colon cancer, the second greatest cancer killer, administration of aspirin and other NSAIDs decreases advanced colon adenomas in Phase II trials and epidemiologic data support their ability to prevent colon cancer. To date prevention trials in the area of lung cancer have shown minimal efficacy. AREAS COVERED The paper examines and discusses in greater detail certain promising agents which the authors have tested either preclinically and or in early phase clinical trials. These agents include RXR agonists, EGFr inhibitors, NSAIDs and Triterpenoids. Other agents including glucocorticoids, pioglitazone and iloprost are briefly mentioned. In addition, the paper presents various types of potential Phase II lung cancer prevention trials and describes their strengths and weaknesses. The potential use of various biomarkers as endpoints in trials e.g. histopathology, non-specific biomarkers (e.g., Ki67, cyclin D expression, apoptosis) and molecular biomarkers (e.g. specific phosphorylated proteins, gene expression etc.) is presented. Finally, we examine at least one approach, the use of aerosols, which may diminish the systemic toxicity associated with certain of these agents. EXPERT OPINION The manuscript presents: a) a number of promising agents which appear applicable to further Phase II prevention trials; b) approaches to defining potential preventive agents as well; c) approaches which might mitigate the side effects associated with potential agents most specifically the use of aerosols. Finally, we discuss biomarker studies both preclinical and clinical which might help support potential Phase II trials. The particular appeal to the preclinical studies is that they can be followed to a tumor endpoint. We hope that this will give the reader further background and allow one to appreciate the potential and some of the hurdles associated with lung cancer chemoprevention.
Collapse
|
37
|
Ma T, Fuld AD, Rigas JR, Hagey AE, Gordon GB, Dmitrovsky E, Dragnev KH. A phase I trial and in vitro studies combining ABT-751 with carboplatin in previously treated non-small cell lung cancer patients. Chemotherapy 2012; 58:321-9. [PMID: 23147218 DOI: 10.1159/000343165] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 09/04/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND ABT-751 is a novel antimitotic agent that exerted cytotoxic effects in preclinical studies. Carboplatin has efficacy in treating advanced non-small cell lung cancer (NSCLC) in combination with other drugs. METHODS Lung cancer cell lines were treated with ABT-751 and/or carboplatin to investigate their impact on cell growth. A phase I study with an expansion cohort was conducted in previously treated NSCLC patients. The primary objective was the maximum tolerated dose (MTD); secondary objectives were objective response rates, median survival, time to tumor progression, dose-limiting toxicities (DLTs), and pharmacodynamic evaluation of buccal swabs. RESULTS Combining ABT-751 with carboplatin significantly reduced growth and induced apoptosis of lung cancer cell lines. Twenty advanced NSCLC patients were enrolled. MTD was ABT-751 125 mg orally twice daily for 7 days with carboplatin AUC 6. DLTs included fatigue, ileus, neutropenia and pneumonitis. Two patients had confirmed partial responses. Median overall survival was 11.7 months (95% CI 5.9-27.0). Time to tumor progression was 2.8 months (95% CI 2.0-2.7). Four of 6 patients showed decreased cyclin D1 protein in posttreatment versus pretreatment buccal swabs. CONCLUSION Combining ABT-751 with carboplatin suppressed growth of lung cancer cell lines and had modest clinical antitumor activity in advanced NSCLC previously treated predominantly with carboplatin. Further studies of this combination are not recommended while investigations of biomarkers in different patient populations, alternative schedules and combinations may be pursued.
Collapse
Affiliation(s)
- Tian Ma
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Galimberti F, Busch AM, Chinyengetere F, Ma T, Sekula D, Memoli VA, Dragnev KH, Liu F, Johnson KC, Guo Y, Freemantle SJ, Andrew AS, Greninger P, Robbins DJ, Settleman J, Benes C, Dmitrovsky E. Response to inhibition of smoothened in diverse epithelial cancer cells that lack smoothened or patched 1 mutations. Int J Oncol 2012; 41:1751-61. [PMID: 22923130 PMCID: PMC3583816 DOI: 10.3892/ijo.2012.1599] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/19/2012] [Indexed: 12/11/2022] Open
Abstract
Hedgehog (HH) pathway Smoothened (Smo) inhibitors are active against Gorlin syndrome-associated basal cell carcinoma (BCC) and medulloblastoma where Patched (Ptch) mutations occur. We interrogated 705 epithelial cancer cell lines for growth response to the Smo inhibitor cyclopamine and for expressed HH pathway-regulated species in a linked genetic database. Ptch and Smo mutations that respectively conferred Smo inhibitor response or resistance were undetected. Previous studies revealed HH pathway activation in lung cancers. Therefore, findings were validated using lung cancer cell lines, transgenic and transplantable murine lung cancer models, and human normal-malignant lung tissue arrays in addition to testing other Smo inhibitors. Cyclopamine sensitivity most significantly correlated with high cyclin E (P=0.000009) and low insulin-like growth factor binding protein 6 (IGFBP6) (P=0.000004) levels. Gli family members were associated with response. Cyclopamine resistance occurred with high GILZ (P=0.002) expression. Newer Smo inhibitors exhibited a pattern of sensitivity similar to cyclopamine. Gain of cyclin E or loss of IGFBP6 in lung cancer cells significantly increased Smo inhibitor response. Cyclin E-driven transgenic lung cancers expressed a gene profile implicating HH pathway activation. Cyclopamine treatment significantly reduced proliferation of murine and human lung cancers. Smo inhibition reduced lung cancer formation in a syngeneic mouse model. In human normal-malignant lung tissue arrays cyclin E, IGFBP6, Gli1 and GILZ were each differentially expressed. Together, these findings indicate that Smo inhibitors should be considered in cancers beyond those with activating HH pathway mutations. This includes tumors that express genes indicating basal HH pathway activation.
Collapse
Affiliation(s)
- Fabrizio Galimberti
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang Y, Rouggly L, You M, Lubet R. Animal models of lung cancer characterization and use for chemoprevention research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 105:211-26. [PMID: 22137433 DOI: 10.1016/b978-0-12-394596-9.00007-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Of the potential sites of cancer development, cancer of the lung accounts for the highest number of cancer deaths each year in the United States (Jemal et al., 2010(1)). Based on its histopathological features, lung cancer is grouped into small cell lung cancer (SCLC; ∼20%) and non-SCLC (NSCLC; ∼80%), which is further divided into three subtypes: squamous cell carcinoma (∼30%), adenocarcinoma (∼50%), and large cell lung carcinoma. Every subtype of lung cancer has a relatively low 5-year survival rate that is attributed, in part, to the fact that they are routinely diagnosed at later histologic stages. Due to this alarming statistic, it is necessary to develop not only new and effective means of treatment but also of prevention. One of the promising approaches is chemoprevention which is the use of synthetic or natural agents to inhibit the initial development of or further progression of early lung lesions (Hong and Sporn, 1997). Many compounds have been identified as potentially effective chemopreventive agents using animal models. Most chemopreventive studies have been performed using mouse models which were developed to study lung adenomas or adenocarcinomas. More recently, models of squamous cell lung cancer and small cell lung cancer have also been developed. This review seeks to highlight mouse models which we helped to develop and presents the results of recent chemopreventive studies that we have performed in models of lung adenocarcinoma, squamous cell carcinoma, and small cell lung cancer.
Collapse
Affiliation(s)
- Yian Wang
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
40
|
Gold KA, Kim ES, Lee JJ, Wistuba II, Farhangfar CJ, Hong WK. The BATTLE to personalize lung cancer prevention through reverse migration. Cancer Prev Res (Phila) 2011; 4:962-72. [PMID: 21733820 DOI: 10.1158/1940-6207.capr-11-0232] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Agents can enter clinical development for cancer prevention either initially or after previous development for a different indication, such as arthritis, with both approaches consuming many years of development before an agent is fully evaluated for cancer prevention. We propose the following, third approach: reverse migration, that is, importing agents, targets, and study designs to personalize interventions and concepts developed in advanced cancer to the setting of cancer prevention. Importing these "ready-made" features from therapy will allow reverse migration to streamline preventive agent development. We recently reported the Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) trial of personalized lung cancer therapy and now propose the reverse migration development of personalized lung cancer prevention based on the BATTLE model.
Collapse
Affiliation(s)
- Kathryn A Gold
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Lung cancer prevention and early detection, which have fallen on hard times for more than the past 20 years, seem to have turned a corner toward better times ahead. Exciting new results of randomized controlled trials that targeted the arachidonic acid pathway, including a celecoxib trial reported by Mao and colleagues in this issue of the journal (beginning on page 984) and a trial of the prostacyclin analog iloprost, complement recently reported 20%-30% lung cancer mortality reductions, either with aspirin in targeting the arachidonic acid pathway or with computed tomography screening. The new results show encouraging activity personalized to former smokers and/or people expressing predictive biomarkers. These trials and technological advances in molecular profiling and imaging herald substantial clinical advances on the horizon of this field.
Collapse
Affiliation(s)
- Fadlo R Khuri
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| |
Collapse
|
42
|
Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 2011; 11:558-72. [PMID: 21734724 DOI: 10.1038/nrc3090] [Citation(s) in RCA: 1030] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cyclin D1, and to a lesser extent the other D-type cyclins, is frequently deregulated in cancer and is a biomarker of cancer phenotype and disease progression. The ability of these cyclins to activate the cyclin-dependent kinases (CDKs) CDK4 and CDK6 is the most extensively documented mechanism for their oncogenic actions and provides an attractive therapeutic target. Is this an effective means of targeting the cyclin D oncogenes, and how might the patient subgroups that are most likely to benefit be identified?
Collapse
Affiliation(s)
- Elizabeth A Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney NSW 2010, Australia
| | | | | | | | | |
Collapse
|
43
|
Kim ES, Lee JJ, Wistuba II. Cotargeting cyclin D1 starts a new chapter in lung cancer prevention and therapy. Cancer Prev Res (Phila) 2011; 4:779-82. [PMID: 21636543 DOI: 10.1158/1940-6207.capr-11-0143] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Lung cancer has limited effective therapy and no effective prevention. Cytotoxic chemotherapy has not improved when combined with the epidermal growth factor receptor (EGFR) inhibitor erlotinib (standard lung cancer therapy) or with the rexinoid bexarotene. Combining erlotinib and bexarotene, however, to cotarget cyclin D1 via the retinoid X receptor and EGFR was active preclinically in KRAS-driven lung cancer cells derived from transgenic mice and in two clinical studies in lung cancer (including wild-type EGFR tumors, with or without KRAS mutations), as reported in this issue of the journal by Dragnev and colleagues (beginning on page 818). These results, along with closely related clinical results of the BATTLE program, support the promise of this cotargeting approach for lung cancer prevention and therapy and of cyclin D1 as a predictive, personalizing marker for it.
Collapse
Affiliation(s)
- Edward S Kim
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | | | | |
Collapse
|